AU2002364692B2 - Methods of screening for toxicity of test compounds - Google Patents

Methods of screening for toxicity of test compounds Download PDF

Info

Publication number
AU2002364692B2
AU2002364692B2 AU2002364692A AU2002364692A AU2002364692B2 AU 2002364692 B2 AU2002364692 B2 AU 2002364692B2 AU 2002364692 A AU2002364692 A AU 2002364692A AU 2002364692 A AU2002364692 A AU 2002364692A AU 2002364692 B2 AU2002364692 B2 AU 2002364692B2
Authority
AU
Australia
Prior art keywords
cell
enzyme
phase
cell line
test compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU2002364692A
Other versions
AU2002364692A1 (en
Inventor
Stephen K Durham
Oliver Flint
Frederic Moulin
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bristol Myers Squibb Co
Original Assignee
Bristol Myers Squibb Co
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bristol Myers Squibb Co filed Critical Bristol Myers Squibb Co
Publication of AU2002364692A1 publication Critical patent/AU2002364692A1/en
Application granted granted Critical
Publication of AU2002364692B2 publication Critical patent/AU2002364692B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5014Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing toxicity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/26Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving oxidoreductase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Urology & Nephrology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Toxicology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • General Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Description

METHODS OF SCREENING FOR TOXICITY OF TEST COMPOUNDS Field of the Invention The invention relates to methods of screening test compounds to determine their effect on cells. In particular, the present invention relates to methods of screening potential drug candidates for toxic effects on human cells.
Background of the Invention Human cell lines have been used in a variety of laboratory applications. For example, immortalized human liver cells have been used in metabolic studies with different chemical classes of carcinogens. Bronchial epithelial cells have been used in studies of the control of squamous differentiation, and identification of chemical and biological agents which induce squamous differentiation. Both of these human cell types have been used in screening chemicals suitable for the treatment of cancer and related diseases, by growing them in vitro in medium containing the chemical to be tested and then, after a suitable period of exposure, determining whether and to what extent genotoxicity, DNA adduct formation, mutagenicity, cell transformation andlor cytotoxicity has occurred following exposure to a carcinogen, by trypan blue exclusion assay or related assays. These cell lines have also been used for identifying agents that induce programmed cell death or apoptosis, which may have an important impact on prevention of malignant transformation. Programmed cell death is assayed by DNA fragmentation or cell-surface antigen analysis.
Human cell lines have also been used to study DNA mutagenesis. Substances known or suspected to be mutagens, or precursors of mutagens, may be added to the growth medium of the cells and then mutations may be assayed, by detection of the appearance of drug resistant mutant cell colonies. Similarly, cell-mediated DNA mutagenesis has been investigated by co-cultivating the cells with cell types known or W¥O 03/050297 PCT/US02/34894 suspected to be capable of secreting mutagenic compounds. Human cell lines have also been used in studies of chromosome damaging agents, studies of malignant transformation, screening for potential chemotherapeutic agents, studies of cellular biochemistry, studies of cellular responses to growth factors and production of growth factors, studies of intracellular communication, characterization of cell surface antigens, hybrid studies for identification of tumor suppressor activity, and identification of novel genes.
In all of the aforementioned studies, the human cell lines used are capable of expressing a cytochrome P450. Cytochromes P450 are a large family of hemoprotein enzymes capable of metabolizing xenobiotics such as drugs, carcinogens and environmental pollutants as well as endobiotics such as steroids, fatty acids and prostaglandins. Some members of the cytochrome P450 family are inducible in both animals and cultured cells, while other constitutive forms are non-inducible. This group of enzymes has both harmful activities, such as the metabolic conversion of xenobiotics to toxic, mutagenic and carcinogenic forms, and beneficial activities, such as the detoxification of xenobiotics.
In the pharmaceutical industry, screening drug candidate compounds for toxicity is a critical step in the drug development process. It is highly desirable to identify, as early as possible, compounds that have an increased likelihood of toxicity.
For example, liver toxicity, such as acute liver failure, is currently the leading cause of drug removal from the market.
Chemical injury to the liver is a multi-faceted phenomenon involving factors such as the nature of the toxic agent, the mechanisms of injury, the nature of the exposure and the susceptibility of the host. A variety of agents can lead to hepatic damage, but in general compounds that are able to injure the liver of most recipients in a variety of species are called "true" or "intrinsic" liver toxicants. Agents that depend on unusual susceptibility of the host to unmask their damaging potential are called "idiosyncratic" hepatotoxins. This increased sensitivity of the host's liver to the damaging effects of the chemical can be linked to two broad categories of mechanisms. The first category is accompanied by clinical and physiological symptoms suggesting the involvement of an immune response and is usually designated as "drug hypersensitivity." The second category consists of liver damage -2appearing in the absence of concomitant immune reaction, and has been called "metabolic idiosyncrasy" (Zimmerman, Hepatoxicity, 2- Lippincott, Williams Wilkins, Philadelphia, PA (1999)). The hypothesis underlying the "metabolic idiosyncrasy" theory is that some products of drug metabolism are responsible for the damage done to the liver cells, but that those metabolites are not produced in sufficient quantities in the majority of the population to result in overt hepatic injury. In some patients, however, the metabolic pathway of the drug favors the production of a toxic species, resulting in liver injury.
There are many obstacles to predicting metabolite-related liver toxicity of pharmaceuticals. Prediction of liver toxicity in clinical trials is poor due to a number of factors, including high polymorphism in the population, differences in metabolism between animal models and humans, and differences in cytochrome P450 expression due to gender and age. In addition, in vitro studies are expensive and time-consuming.
One of the major obstacles in the drug development process is predicting 15 which drug candidates will exhibit idiosyncratic toxicity. A major challenge in predicting metabolic idiosyncrasy is not to find models that are representative of the majority of the population, but rather to find models that are representative of those few subjects for which the metabolism is oriented toward toxicity. As compared to intrinsic toxicity, idiosyncratic toxicity is far more difficult to predict, due to the nature of the toxicity. Intrinsic toxicity is: typically a property of the drug; a constant occurrence; dose-dependent; characterized by a clearonset of toxicity; and predicted by animal models. Idiosyncratic toxicity, on the other hand, is: typically a result of interaction between the drug and patient specific factors; a relatively rare occurrence (<1/10,000); usually independent of dose; characterized by a delayed onset; and difficult (if not impossible) to predict with animal models.
Thus, there is a need for rapid, affordable, accurate methods of screening compounds for toxicity. Specifically, there is a need for methods to predict idiosyncratic toxicity of a drug candidate. In particular, there is a need for methods to identify idiosyncratic toxicity early in the drug development process. There is also a need for methods of screening compounds for toxicity which reflect human metabolism and are representative of the diverse human population.
The discussion of documents, acts, materials, devices, articles and the like is included in this specification solely for the purpose of providing a context for the present invention. It is not suggested or represented that any or all of these matters formed part of the prior art base or were common general knowledge in the field relevant to the present invention as it existed in Australia before the priority date of each claim of this application.
Throughout the description and claims of the specification, the word "comprise" and variations of the word, such as "comprising" and "comprises", is not intended to exclude other additives, components, integers or steps.
IDSummary of the Invention The present invention features methods of screening a compound to determine the toxic effect of the compound on cells. In one aspect, the invention provides a method of O determining whether a compound exhibits idiosyncratic toxicity by contacting a test compound S 5 with a cell which expresses a Phase I enzyme, contacting the cell with a cell viability indicator to IND determine whether the test compound has generated a toxic metabolite, wherein generation of a toxic metabolite indicates that the test compound exhibits idiosyncratic toxicity.
In accordance with one aspect, the present invention provides a method of determining whether a compound exhibits idiosyncratic toxicity comprising: a) contacting a test compound with at least one first cell line which expresses a Phase I IDenzyme and with a second cell line which does not express a Phase I enzyme; (Ni b) contacting the first and second cell lines with a cell viability indicator to determine whether the first cell line has generated a toxic metabolite from said test compound; and c) observing for any reduction in cell viability of the first cell line relative to the second cell line, wherein a greater reduction of cell viability of the first cell line relative to the cell viability of the second cell line is indicative of the generation of the toxic metabolite and that the test compound exhibits idiosyncratic toxicity.
In another aspect, the present invention provides a method of determining whether a compound exhibits idiosyncratic toxicity comprising: a) contacting a test compound suspected of having idiosyncratic toxicity with a first cell line which expresses a cytochrome P-450 enzyme and with a second cell line which does not express a cytochrome P-450 enzyme.
b) contacting the first and second cell lines with a cell viability indicator to determine whether the first cell line has generated a toxic metabolic from said test compound; and c) observing for any reduction in cell viability of the first cell line relative to the second cell line, wherein a greater reduction of cell viability of the first cell line relative of the cell viability of the second cell line is indicative of the generation of the toxic metabolite and that the test compound exhibits idiosyncratic toxicity.
In a further aspect, the present invention provides a method of determining idiosyncratic toxicity of a compound comprising: a) contacting a test compound suspected of having idiosyncratic toxicity with a plurality of first cell lines and with a second cell line which does not express a Phase I enzyme each first cell line expressing a difference Phase I enzyme.
b) contacting the first and second cell lines with a cell viability indicator to determine whether the first cell line has generated a toxic metabolite from said test compound; and c) observing for any reduction of cell viability in any of the first cell lines relative to the second cell line is indicative of the generation of the toxic metabolite and that the test compound exhibits idiosyncratic toxicity.
The present invention also provides a method for determining the likelihood of hepatic liability in a population following administration of a test compound comprising: Yv:\ouise\SeMS sXasl76O97 _spec dean. 02 1l.06.doc I0 a) contacting a test compound with a plurality of first cell lines, each first cell line
O
0expressing a different cytochrome P-450 enzyme, and with a second cell lien which does not express a cytochrome P-450 enzyme.
O b) contacting the first and second cell lines with a cell viability indicator to determine whether the first cell has generated a toxic metabolite from said test compound; and I c) observing for any changes in viability of the first cell lines relative to the viability of the second cell line, wherein greater cell viability found for any of the first cell lines relative to the second cell line is indicative that a population deficient in the cytochrome P-450 enzyme expressed by any of the firsts cell lines having greater cell viability after test compound contact has a likelihood of hepatic liability following administration of the test compound to the IND population.
In a preferred embodiment, the indicator is a chromogenic compound, more preferably, 0 a tertazolium compound, and most preferably, the indicator is3- 5-dimethylthiazol-2-yl)-5- (3carboxymethoxyphenyl)-2- (4-sulfophenyl)-2H-tetrazolium, inner salt). The most preferred cell line used in the method of the invention is theTHLE-5B cell line (also referred to Preferred cytochromes P450 include 3A4,2C9, 2C19, 2D6,IA1, 1A2, 2B6,2C11 and 2E1. Of these, 3A4,2C9,2C19, and 2D6 are the most preferred cytochromes P450 for use with the invention.
The methods of the invention may be performed manually. Preferably, the methods of the invention are performed using an automated system to achieve high throughput.
Brief Description of the Drawings Figure 1 is a graphical representation ofmRNA expression levels in THLE-5 cells for key Phase I (left) and Phase II (right) enzymes, as compared to primary human hepatocytes (PHH).
Figure 2 is a graphical comparison of the metabolism of buspirone by human liver microsomes, 3A4Supersomeso, 3A4 and TC5 cell lines.
Figure 3 shows the results for control samples used in a high throughput assay of the invention.
Figure 4 depicts sample data obtained from a method of the invention.
Figure 5 demonstrates reduced toxicity of a parent compound by CYP2C19 metabolism.
Figure 6 shows increased toxicity following CYP2C19 metabolism.
Y:UOuSe\EMSXSpoeSX716897-Speal clean. 02.1 1.06.dO WO 03/050297 PCT/US02/34894 Detailed Description of the Invention In one aspect the present invention takes advantage of the metabolization of chemical compounds by liver cells. Generally speaking, the elimination of a chemical compound by the liver usually involves two steps. In the first step, some portion of the compound's chemical structure is modified to form a "chemically-reactive" end.
In the second step, a large, hydrophilic group is added to that reactive portion to disrupt the three-dimensional structure of the parent compound and facilitate its elimination in biological fluids. These steps form the two "phases" of drug metabolism, and are each catalyzed by specific families of enzymes. Cytochromes P450 are among the enzymes responsible for "Phase I" reactions, or the formation of a reactive entity.
In some cases, there may be a desynchronization between the production of reactive species by the phase one enzyme reactions, and subsequent elimination by conjugation during phase two. For example, desynchronization occurs when there is an increase in the amount of Phase I enzymes, causing an increase in reactive metabolites, but no corresponding increase in Phase HT enzymes. Alternatively, desynchronization may result from an absence or depletion of Phase II enzymes, such as results from polymorphism. However, depletion of Phase II enzymes may also occur in subjects who are treated with an additional substance, such as a bloodthinning compound or an herbal supplement. In these subjects, metabolites of the additional substance may compete for the Phase II enzyme that otherwise would be present to act on the reactive metabolites formed by the Phase I enzymes. As a result of desynchronization, reactive species may accumulate inside the cytoplasm and may bind to macromolecules, proteins, involved in cell structure or metabolism. The alteration of these macromolecules may result in the loss of critical functions and cellular toxicity.
In one embodiment, the present invention provides a method of screening chemical compounds for toxicity by increasing the amount of Phase I enzymes of a specific type, while taking advantage of a reduction in the Phase n1 enzyme production in the host cells (See Figure In a preferred embodiment, the amount of Phase I enzymes in a cell is increased through the use of an expression vector. According to the method of the invention, the transfected cells have a vastly increased potential to WO 03/050297 PCT/US02/34894 generate reactive metabolites directly in their cytoplasm and demonstrate any toxic effect. In this manner, the method of the invention simulates conditions that may arise in the liver of individuals who are susceptible to idiosyncratic toxicity. In a preferred embodiment, the method of the invention includes a cell having increased expression levels of a Phase I enzyme. For example, increased expression of a Phase I enzyme is at least about 20 fold more, more preferably at least about 50 fold more, and most preferably, at least about 100 fold more than expression of the same enzyme in primary human hepatocytes (PHH). In another preferred embodiment, the method of the invention includes a cell having decreased expression of a Phase II enzyme. For example, decreased expression of a Phase II enzyme is at least about 70%, more preferably at least about 80%, and most preferably, at least about 90% less than expression of the same enzyme in PHH.
In general, any cell line that expresses or is made to express a cytochrome P450 may be used in the invention. Cell lines derived from human liver parenchymal cells are preferred for the prediction of hepatotoxicity. Immortalized human cell lines for use in the method of the invention may be obtained through methods known in the art. For example, human hepatocyte cell lines may be immortalized by transformation with simian virus 40 large T antigen, as described in Pfeifer, et al., "Simian virus 40 large tumor antigen-immortalized normal human liver epithelial cells express hepatocyte characteristics and metabolize chemical carcinogens," Proc. Natl. Acad.
Sci. U.S.A. 90, 5123-5127 (1993), hereby incorporated by reference in its entirety.
The immortalized human cells are reported to be similar to quiescent normal human hepatocytes, are non-tumorigenic when injected into nude mice, have near-diploid karyotypes, do not express alpha-fetoprotein, and are free of Hepatitis B or human immunodeficiency virus (Pfeifer et al, 1993). The THLE-5 cell line (also sometimes referred to as "THLE-5B") was deposited under the terms and conditions of the Budapest Treaty at the American Type Culture Collection on Apr. 23, 1992, and was assigned the accession number CRL11113. The parent cell line, THLE-5B-cl5 c 15) does not express Phase I metabolizing activity, but retains normal expression of most Phase II metabolizing enzymes, such as glutathione S-transferase, epoxide hydrolase, N-acetyl transferase type I, aldehyde reductase, and quinone reductase, as well as detoxification systems, such as superoxide dismutase (SOD) and catalase.
WO 03/050297 PCT/US02/34894 In a preferred embodiment, multiple cells, each expressing a different cytochrome P450, are used in the method of the invention. Four separate cell lines, THLE-5B-3A4, THLE-5B-2C9, THLE-5B-2C19, and THLE-5B-2D6, expressing the different cytochrome P450s indicated by their names (3A4, 2C9, 2C19, and 2D6, respectively), were derived by transfection of THLE-5B-cl5 with a plasmid containing a CMV expression vector, human cDNA coding for the specific P450 isoenzyme and a selection marker, using methods known in the art.
The use of these four cell lines allows for the determination of the specific role of each of these enzymes in the generation of toxic metabolites or detoxification of a parent compound. Thus, in another embodiment of the present invention, information gathered by this method of the invention may be used to predict the likelihood of toxicity in particular population groups. For example, detoxification of a parent compound by CYP 2D6 only may be shown by a decrease in cell viability with increased drug concentration in the parent TC5 cell line and three other daughter lines, but not in the 2D6 cell line. These results suggest, in a population that is deficient in this enzyme, an increased likelihood of hepatic liability.
The invention is based, in part, on the effect cytochrome P450 polymorphism can have on drug metabolism. Six isoenzymes of cytochrome P450 (2E1, 2C19, 2C9, 3A4, 2D6, 1Al, and 1A2) account for about 95% of all cytochrome P450-mediated reactions. Of these, 2D6, 2C9, and 2C19 are the cytochromes P450 that account for the majority of polymorphism associated with clinically relevant changes in drug effects. See Evans, W.E. and Relling, "Pharmacogenomics: translating functional genomics into rational therapeutics," Science 286(5439): 487-91 (Oct. 1999).
FIG. 1 depicts mRNA expression levels for key Phase I (left) and Phase II (right) enzymes, as compared to primary human hepatocytes (PHH). In FIG. 1, the level of expression of the metabolism enzymes have been normalized to the level of expression of the same enzyme in primary human hepatocytes. As shown on the left graph, the cells express many fold more mRNA of the respective P450 than primary human hepatocytes. In addition, phase II enzymes, such as UDPGT, are approximately 10 fold less expressed. This differential expression enhances the formation of potential reactive metabolites as well as their ability to react with critical -7- WO 03/050297 PCT/US02/34894 cellular components.
FIG. 2 compares the metabolism of buspirone by human liver microsomes, 3A4 Supersomes®, 3A4, and TC5 cell lines. The metabolic profile of the 3A4 cell line closely matches the results obtained with microsomes extracted from human livers or insect cell line, demonstrating that metabolites produced in the testing system are very likely to be also produced in a subject's liver. As expected, the parent cell line does not show any metabolism of the test compound even after 44 hours incubation.
FIG. 3 shows the results for control samples used in a high throughput assay of the invention. Cell-only and media-only controls may be included on every plate. In addition, two compounds, perhexiline (positive control of toxicity known human liver toxicant) and theophylline (negative control of toxicity) may be included in each test. The interplate variation coefficient is calculated from cell-only and media-only wells present on each plate.
FIG. 4 depicts sample data obtained from a method of the invention. The compound on the left exhibited toxicity in the TC5 cell line as well as all the transfected cell lines. It is therefore predicted to be an intrinsic liver toxicant. The compound on the right is not predicted to have increased likelihood of clinical liver liability.
FIG. 5 demonstrates reduced toxicity of a parent compound by CYP 2C19 metabolism. The assay shows a marked amelioration of the parent compound toxicity following metabolism by cytochrome P450 2C19. This compound might be harmless for a majority of the population. However, increased liver toxicity may appear in the subpopulation of poor CYP 2C19 metabolizers.
FIG. 6 demonstrates increased toxicity following CYP 2C19 metabolism. In such a case, it might be helpful to test the compound on primary human hepatocytes to assess the importance of the 2C19 route in the overall metabolism of the drug candidate.
The following solutions and reagents were prepared and used in the experiments described herein.
Culture Medium (CM) Low calcium medium, PMFR-4, supplemented with 2 mM L-glutamine, -8- WO 03/050297 PCT/US02/34894 gg/mL gentamycin, 1.75 iM insulin, 0.2 gM hydrocortisone, 5 ng/mL epidcrmal growth factor, 10 gg/mL transferrin, 500 nM phosphoryethanolamine/ethanolamine, nM triiodothyronine, 15 gg/mL bovine pituitary extract, 0.33 nM retinoic acid, and 3% fetal bovine serum (Biofluids).
Trypsin EDTA IX Life Technologies.
Solutions of 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4sulfophenyl)-2H-tetrazolium, inner salt and phenazine methosulfate (PMS) were prepared as follows. Because the MTS solution is light sensitive, it was prepared fresh for each run of the assay.
MTS Solution A 2 mg/mL MTS solution was prepared by dissolving 240 mg MTS (Promega Corp.) powder in 120 mL of Dulbecco's PBS (Ca 2 Mg2+ free, Life Technologies). The solution was warmed as necessary to dissolve the powder, then filtered through a 0.22 pm filter.
PMS Solution A 0.92 mg/mL solution of phenazine methosulfate (PMS, Sigma) in PBS was prepared and filtered through a 0.22 pm filter.
The two solutions are combined at a ratio of 20ml of MTS to 1ml of PMS for use.
Coating Media To 500 mL of LHC basal medium (Biofluids, #118) was added human fibronectin 5 mg (Collaborative Biomedical Products, #40008A), Vitrogen 100 (purified bovine collagen) 5 mL (Collagen Corp #PC0701), BSA stock (Biofluids #343). The solution was filtered through a 0.22 gm filter.
A sufficient amount of coating medium is added to cover the well or flask, followed by incubation for at least for 15 minutes at 37 0 C. Coating media was removed prior to seeding flask or well.
Cell Preparation and Culture Cells were grown at 37 0 C, under 5% CO 2 The cells were serially passaged in 175cm 2 culture flasks, split 1:10, every 7 days. Approximately every 5th passage, cells were selected by addition of 150 Rig/ml of G418. Periodically, metabolic activity of the daughter lines were verified by usage of P450-specific fluorescent substrates.
-9- WO 03/050297 PCT/US02/34894 For cell passage, the cells were rinsed with PBS, incubated 5 minutes with trypsin (trypsin EDTA IX Life Technologies), and harvested in culture medium.
For experimental treatments, cells were seeded at 1.5 x 10 4 cells/well in 96-well culture dishes (Falcon) in a 100 pL volume.
Test Compound Preparation and Administration All test compounds were dissolved in DMSO at 25 mg/mL (25,000 gg/mL) and 5 mg/mL (5,000 gg/mL), which was added to the culture medium to a final concentration of 1% Before testing, starting concentrations of 250 and gg/mL were read in a SPECTRAmaxTM spectrophotometer at 450 nm to evaluate the solubility. If a compound was not soluble at 50 gpg/mL, serial dilutions were prepared and tested to determine the highest soluble concentration. For testing in the robotic system, the initial concentration in the mother plate was 250 jgg/mL or 50 gg/mL (2X concentration). These two starting concentrations will produce overlapping curves.
The first dilution and highest concentration tested was 125 pg /mL or 25 gg /mL in 0.5% DMSO and the final volume was 100 gL. The further serial dilutions were 2fold, 62.5, 31.25, 15.6, and 7.8 ptg/mL (12.5, 6.25, 3.125, 1.625 jtg/mL) and the DMSO was diluted 2-fold in the same pattern. Culture medium containing test compound (100 [tL) was added to the cells after a confluent monolayer had formed.
Positive perhexiline) and negative theophylline) reference agents were included in each test.
In the automated or "robotic" procedure, a "mother plate" (96-well) containing test articles in triplicate at twice the highest concentration to be tested (25 mg/mL), served as the supply of compounds for the assay. From this mother plate, five successive 2-fold serial dilutions (100 |tL) were made, using the robotic arm (CRS Robotics, Burlington, Ontario) and Quadra pipettor for each cell line. For example, a compound made at a concentration of 200 [AM into the mother plate would be tested on the cells in plate 1 at 100 [tM, at 50 jiM in plate 2, at 25 tM in plate 3, at 12.5 [tM in plate 4 and at 6.25 jtM in plate 5. These 5 different concentrations would be repeated for each of the 5 cell lines, for a total of 25 different plates. The plates were incubated in a Hotpack incubator at 37 oC under 5% CO 2 for 20 hrs. Following the incubation period, 22 pL of the combined PMS-MTS solution was added using the WO 03/050297 PCT/US02/34894 robotic arm and Quadra pipettor. The test plates were again transferred to the incubator. After an additional 2 hr incubation with MTS, the plates were individually transported to a plate reader (Wallac Victor 1420) and absorbance intensity was measured at 490 nm.
In the manual procedure, test compounds at twice the highest concentration to be tested, served as the supply of compounds for the assay. From this mother plate, five successive 2-fold serial dilutions were made for each cell line. For example, a compound made at a concentration of 200[M into the mother plate would be tested on the cells in plate 1 at 100 at 50 in plate 2, at 25 [tM in plate 3, at 12.5 [tM in plate 4 and at 6.25 p.M in plate 5. These 5 different concentrations are repeated for each of the 5 cell lines, for a total of 25 different plates. A hand-held multi-pipettor was used to perform the steps in the manual assay. The plates were incubated at 37 °C under 5% CO 2 for 20 hrs in a standard laboratory incubator. Following the incubation period, 22 gL of the combined PMS-MTS solution were added to each well. After an additional 2 hr incubation, absorbance intensity was measured at 490 nm using a Wallac Victor 1420 plate reader.
Cell Viability Assessment MTS is an indicator of mitochondrial function. The assay is thus a colorimetric method for determining the number of viable cells in proliferation. MTS, in the presence of an electron coupling reagent (phenazine methosulfate), is bioreduced by cells into a formazan that is soluble in tissue culture medium. The conversion of MTS into the aqueous soluble formazan is accomplished by dehydrogenase enzymes found in metabolically active cells. The quantity of formazan product as measured by the amount of absorbance at 490 nm is directly proportional to the number of metabolically active cells in culture.
Analysis of Results Validation of the run was made by comparing three criteria (perhexiline IC 5 o, theophylline IC 5 o and plate-to-plate coefficient of variation) with historical references.
If a test was rejected, then all data from this particular run were discarded and all test articles were rerun. Data from validated tests were adjusted for background absorbance (defined by the "medium-only wells) as well as plate-to-plate variability, expressed as percentage of the viability observed in the cell-only wells and examined -11 WO 03/050297 PCT/US02/34894 for significant concentration-related changes with respect to the endpoint measured.
For each replicate of the test compound, concentration-response curves were plotted for the different cell lines and an IC 5 0 determined using a four-parameters logistic regression model. The IC 5 0 is defined as the concentration corresponding to a viability on the regression line. A mean and standard deviation (SD) is calculated from the three IC50 obtained from the replicates. These data are reported for each test compound, as well as positive and negative controls.
Interpretation of Results For compounds with maximum solubility greater than 50 uM.
Test compounds with an IC 50 in any of the cell lines less than 50+SD were predicted to have an increased likelihood of clinical hepatic liabilities. Test compounds with an IC 5 0 in all cell lines greater than 50+SD jiM were predicted to have a reduced likelihood of clinical hepatic liabilities.
For compounds with maximum solubility less than 50 i.M Test compounds with an IC 50 in any of the cell lines were predicted to have an increased likelihood of clinical hepatic liabilities. If the dose response curve allows extrapolation to 50 test articles with an extrapolated IC5o in all cell lines greater than 50+SD giM were predicted to have a reduced likelihood of clinical hepatic liabilities and test articles with an extrapolated ICso less than 50 itM in any of the cell lines were predicted to have an increased likelihood of clinical hepatic liabilities. If the maximum solubility reached in the assay does not allow extrapolation to 50 [tM, the likelihood of clinical hepatic liabilities cannot be predicted.
EXAMPLES
Example 1 Immortalized human hepatocytes were plated at 1.5 x10 4 cells per well in 100 I.L culture media on coated plates. These cells were incubated overnight at 37 °C under 5% CO 2 A confluent monolayer formed during this incubation. Cells were exposed to test compounds by adding 100 pL of test compound to cells at 2X the concentration to be tested. Serial dilutions were made for lower concentrations (1 concentration per plate). Cells were then incubated at 37 OC under 5% CO 2 for 20 hrs.
-12- WO 03/050297 PCT/US02/34894 MTS reagent was added in 22 pL volume to each well. Cells were then incubated for 2 hrs, and the absorbance at 490 nm was determined.
All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each independent publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
13-

Claims (12)

1. A method of determining whether a compound exhibits idiosyncratic toxicity comprising: a) contacting a test compound with at least one first cell line which expresses a Phase I O enzyme and with a second cell line which does not express a Phase I enzyme; Z 5 b) contacting the first and second cell lines with a cell viability indicator to determine IN whether the first cell line has generated a toxic metabolite from said test compound; and c) observing for any reduction in cell viability of the first cell line relative to the second cell line, wherein a greater reduction of cell viability of the first cell line relative to the cell viability of the second cell line is indicative of the generation of the toxic metabolite and that the test 10 compound exhibits idiosyncratic toxicity. (N 2. The method of claim 1, wherein the first cell line is THLE-5B-3A4, THLE-5B-2C9, THLE- S5B-2CI9, or THLE-5B-2D6.
3. The method of claim 1 or claim 2, wherein the Phase I enzyme is a eytochrome P450.
4. The method of claim 3, wherein the cytochrome P450 is 3A4, 2C9, 2C 19, 2D6, 1AI, 1A2,2B6, 2Cll or 2EI.
5. The method of any one of claims 1 to 4, further comprising a plurality of cells, wherein each cell expresses a different Phase I enzyme.
6. The method of claim 5, wherein the plurality of cells comprises THLE-5B- 3A4, THLE- 5B-2C9, THLE-5B-2C19, and THLE-5B-2D6.
7. The method of any one of claims 1 to 6, wherein the indicator is 3- 2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt.
8. The method of any one of claims 1 to 7, wherein the cell expresses the Phase I enzyme at a level that is at least about 20 fold more than the level of expression of the Phase I enzyme in primary human hepatocytes. Y:%LourseBMs sSpeie\1 71887-gp e ldean. 02.1l.O.doc \O
9. The method of any one of claims 1 to 9, wherein the cell expresses the Phase I enzyme O at a level that is at least about 50 fold more than the level of expression of the Phase I enzyme S 5 in primary human hepatocytes. \O The method of claim 1, wherein the cell expresses the Phase I enzyme at a level that is at least about 100 fold more than the level of expression of the Phase I enzyme in primary \human hepatocytes. ID11. The method of any one of claims 1 to 10, wherein the cell expresses a Phase II enzyme at a level that is at least about 70% less than the level of expression of the Phase II enzyme in primary human hepatocytes.
12. The method of any one of claims 1 to 11, wherein the cell expresses a Phase II enzyme at a level that is at least about 80% less than the level of expression of the Phase II enzyme in primary human hepatocytes.
13. The method of any one of claims 1 to 12, wherein the cell expresses a Phase II enzyme at a level that is at least about 90% less than the level of expression of the Phase II enzyme in primary human hepatocytes.
14. A method of determining whether a compound exhibits idiosyncratic toxicity comprising: a) contacting a test compound suspected of having idiosyncratic toxicity with a first cell line which expresses a cytochrome P-450 enzyme and with a second cell line which does not express a cytochrome P-450 enzyme. b) contacting the first and second cell lines with a cell viability indicator to determine whether the first cell line has generated a toxic metabolic from said test compound; and c) observing for any reduction in cell viability of the first cell line relative to the second cell line, wherein a greater reduction of cell viability of the first cell line relative of the cell viability of the second cell line is indicative of the generation of the toxic metabolite and that the test compound exhibits idiosyncratic toxicity. vYU oLoBMS Spocde71M697_speci. dee 02.11.06dc 16 \O A method of determining idiosyncratic toxicity of a compound comprising: a) contacting a test compound suspected of having idiosyncratic toxicity with a plurality O of first cell lines and with a second cell line which does not express a Phase I enzyme each first S 5 cell line expressing a difference Phase I enzyme. IND b) contacting the first and second cell lines with a cell viability indicator to determine whether the first cell line has generated a toxic metabolite from said test compound; and c) observing for any reduction of cell viability in any of the first cell lines relative to the second cell line is indicative of the generation of the toxic metabolite and that the test compound exhibits idiosyncratic toxicity. S16. A method for determining the likelihood of hepatic liability in a population following administration of a test compound comprising: a) contacting a test compound with a plurality of first cell lines, each first cell line expressing a different cytochrome P-450 enzyme, and with a second cell lien which does not express a cytochrome P-450 enzyme. b) contacting the first and second cell lines with a cell viability indicator to determine whether the first cell has generated a toxic metabolite from said test compound; and c) observing for any changes in viability of the first cell lines relative to the viability of the second cell line, wherein greater cell viability found for any of the first cell lines relative to the second cell line is indicative that a population deficient in the cytochrome P-450 enzyme expressed by any of the firsts cell lines having greater cell viability after test compound contact has a likelihood of hepatic liability following administration of the test compound to the population.
17. A method according to any one of claims 1, 14, 15 or 16 substantially as hereinbefore described with reference to the figures. Y:'LowsevBMSXSpeoes%718O97_sped. dean. 02.11.06 Coc
AU2002364692A 2001-10-31 2002-10-31 Methods of screening for toxicity of test compounds Ceased AU2002364692B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US33650901P 2001-10-31 2001-10-31
US60/336,509 2001-10-31
PCT/US2002/034894 WO2003050297A1 (en) 2001-10-31 2002-10-31 Methods of screening for toxicity of test compounds

Publications (2)

Publication Number Publication Date
AU2002364692A1 AU2002364692A1 (en) 2003-06-23
AU2002364692B2 true AU2002364692B2 (en) 2006-11-23

Family

ID=23316423

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2002364692A Ceased AU2002364692B2 (en) 2001-10-31 2002-10-31 Methods of screening for toxicity of test compounds

Country Status (8)

Country Link
US (1) US7041501B2 (en)
EP (1) EP1446497A4 (en)
JP (1) JP2005511093A (en)
AU (1) AU2002364692B2 (en)
CA (1) CA2465262A1 (en)
IL (1) IL161300A0 (en)
MX (1) MXPA04004018A (en)
WO (1) WO2003050297A1 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7759113B2 (en) * 1999-04-30 2010-07-20 The General Hospital Corporation Fabrication of tissue lamina using microfabricated two-dimensional molds
US7776021B2 (en) * 2000-04-28 2010-08-17 The Charles Stark Draper Laboratory Micromachined bilayer unit for filtration of small molecules
WO2004065616A2 (en) 2003-01-16 2004-08-05 The General Hospital Corporation Use of three-dimensional microfabricated tissue engineered systems for pharmacologic applications
EP1636351A4 (en) 2003-05-21 2007-04-04 Gen Hospital Corp Microfabricated compositions and processes for engineering tissues containing multiple cell types
US7452678B2 (en) * 2003-06-24 2008-11-18 Bristol-Myers Squibb Company Identification of biomarkers for liver toxicity
US7235353B2 (en) * 2003-07-18 2007-06-26 Cytokinetics, Inc. Predicting hepatotoxicity using cell based assays
US20050014217A1 (en) * 2003-07-18 2005-01-20 Cytokinetics, Inc. Predicting hepatotoxicity using cell based assays
WO2007146203A1 (en) * 2006-06-08 2007-12-21 The University Of North Carolina At Chapel Hill Pulsing of bile compartments in sandwich-cultured hepatocytes
WO2012031057A1 (en) 2010-09-01 2012-03-08 Bristol-Myers Squibb Company Bms- 582949 for the treatment of resistant rheumatic disease
JP2017035081A (en) * 2015-08-13 2017-02-16 サントリーホールディングス株式会社 Test method of animal cell toxicity or antioxidant ability

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5164313A (en) 1987-06-05 1992-11-17 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Recombinant vaccinia virus encoding cytochromes P-450
US5356806A (en) 1987-06-05 1994-10-18 The United States Of America As Represented By The Department Of Health And Human Services Immortalized human cell lines containing exogenous cytochrome P450
US5506131A (en) 1987-06-05 1996-04-09 The United States Of America As Represented By The Department Of Health And Human Services Immortalized human cell lines containing exogenous cytochrome P450 genes
US5665589A (en) 1988-12-14 1997-09-09 The United States Of America As Represented By The Department Of Health And Human Services Human liver epithelial cell lines
WO1990007007A1 (en) 1988-12-14 1990-06-28 The United States Of America, Represented By The Secretary, United States Department Of Commerce Cell culture medium for human liver epithelial cell line
US5529920A (en) 1988-12-14 1996-06-25 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Human liver epithelial cell line and culture media therefor
JPH0749923B2 (en) * 1989-05-31 1995-05-31 大日本スクリーン製造株式会社 Unit converter for laser length measuring equipment
US5457187A (en) * 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
US5759782A (en) * 1995-06-07 1998-06-02 The United States Of America Cellular apoptosis susceptibility protein (CSP) and antisense CSP
US5869243A (en) 1996-03-05 1999-02-09 Rhode Island Hospital Immortalized hepatocytes
TW471968B (en) * 1999-08-25 2002-01-11 Committee On Chinese Medicine Solamargine pharmaceutical composition for killing cancer cells
US6852845B1 (en) * 1999-11-22 2005-02-08 Curagen Corporation Method of identifying toxic agents using NSAID-induced differential gene expression in liver
US7700822B2 (en) * 2002-07-12 2010-04-20 Cancer Research Technology Limited Modulation of cytochrome P450 reductase activity

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
References cited in WO 03/050297 *

Also Published As

Publication number Publication date
CA2465262A1 (en) 2003-06-19
IL161300A0 (en) 2004-09-27
MXPA04004018A (en) 2004-07-23
AU2002364692A1 (en) 2003-06-23
US20030113708A1 (en) 2003-06-19
WO2003050297A1 (en) 2003-06-19
JP2005511093A (en) 2005-04-28
EP1446497A1 (en) 2004-08-18
EP1446497A4 (en) 2004-12-29
US7041501B2 (en) 2006-05-09

Similar Documents

Publication Publication Date Title
Bell et al. Comparison of hepatic 2D sandwich cultures and 3D spheroids for long-term toxicity applications: a multicenter study
Roth et al. Idiosyncratic Drug‐Induced Liver Injury (IDILI): Potential Mechanisms and Predictive Assays
AU2002364692B2 (en) Methods of screening for toxicity of test compounds
Braunbeck et al. Fish ecotoxicology
Cox et al. The utility of metabolic activation mixtures containing human hepatic post-mitochondrial supernatant (S9) for in vitro genetic toxicity assessment
Ware et al. Exploring chronic drug effects on microengineered human liver cultures using global gene expression profiling
EP0700442B1 (en) Immortalized human cell lines containing exogenous cytochrome p450 genes
Seo et al. Performance of high-throughput CometChip assay using primary human hepatocytes: a comparison of DNA damage responses with in vitro human hepatoma cell lines
Jetten et al. Interindividual variation in response to xenobiotic exposure established in precision-cut human liver slices
Dörrenhaus et al. Induction of unscheduled DNA synthesis in primary human urothelial cells by the mycotoxin ochratoxin A
Salazar et al. Relationship between micronuclei formation and p53 induction
Strickland et al. Biomarkers for assessing environmental exposure to carcinogens in the diet
Müller-Tegethoff et al. Application of the in vitro rat hepatocyte micronucleus assay in genetic toxicology testing
Brendt et al. Is a liver comparable to a liver? A comparison of different rat-derived S9-fractions with a biotechnological animal-free alternative in the Ames fluctuation assay
Mondal et al. High-content micronucleus assay in genotoxicity profiling: initial-stage development and some applications in the investigative/lead-finding studies in drug discovery
Lemaire et al. Toxicity assays in nanodrops combining bioassay and morphometric endpoints
Jones et al. 6 Cytochrome P450 Metabolism and Inhibition: Analysis for Drug Discovery
Billis et al. High‐content imaging assays for identifying compounds that generate superoxide and impair mitochondrial membrane potential in adherent eukaryotic cells
Cox Evaluation of strategies to improve in vitro mutagenicity assessment: Alternative sources of S9 exogenous metabolic activation and the development of an in vitro assay based on MutaMouse primary hepatocytes
Kopp et al. Liver-on-chip model and application in predictive genotoxicity and mutagenicity of drugs
Farrell et al. High-throughput Screening of Toxic Chemicals on Neural Stem Cells
Rodrigues et al. Measurement of cytochrome P450 enzyme induction and inhibition in human hepatoma cells
Boisvert Critical Examination of Selected Aspects of the ToxTracker In Vitro Genotoxicity Assay: Evaluation of S9 Metabolic Activation Protocols and Quantitative Interpretation of Dose-response Data
Van Poppel et al. Biomarkers in epidemiological and toxicological nutrition research
Gudi et al. Genetic Toxicology Studies

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired